REBLOZYL Powder for solution for injection Ref.[10651] Active ingredients: Luspatercept

Source: European Medicines Agency (EU)  Revision Year: 2020 

5.1. Pharmacodynamic properties

Pharmacotherapeutic group: Antianaemic preparations, other antianaemic preparations
ATC code: B03XA06

Mechanism of action

Luspatercept, an erythroid maturation agent, is a recombinant fusion protein that binds selected transforming growth factor-β (TGF-β) superfamily ligands. By binding to specific endogenous ligands (e.g. GDF-11, activin B) luspatercept inhibits Smad2/3 signalling, resulting in erythroid maturation through differentiation of late-stage erythroid precursors (normoblasts) in the bone marrow. Smad2/3 signalling is abnormally high in disease models characterised by ineffective erythropoiesis, i.e. MDS and β-thalassaemia, and in the bone marrow of MDS patients.

Clinical efficacy and safety

Myelodysplastic syndromes

The efficacy and safety of luspatercept were evaluated in a Phase 3 multicentre, randomised, double-blind, placebo-controlled study MEDALIST (ACE-536-MDS-001) in adult patients with anaemia requiring RBC transfusions (≥2 units/8 weeks) due to International Prognostic Scoring System-Revised (IPSS-R) very low-, low- or intermediate-risk MDS who have ring sideroblasts (≥15%). Patients were required to have either received prior treatment with an erythropoiesisstimulating agent (ESA) with inadequate response, to be ineligible for ESAs (determined to be unlikely to respond to ESA treatment with serum erythropoietin (EPO) >200 U/L), or intolerant to ESA treatment. Patients with deletion 5q (del5q) MDS were excluded from the study.

Patients in both arms were treated for 24 weeks, then continued treatment if they had demonstrated clinical benefit and absence of disease progression. The study was unblinded for analyses when all patients had at least received 48 weeks of treatment or discontinued treatment.

A total of 229 patients were randomised to receive luspatercept 1.0 mg/kg (n=153) or placebo (n=76) subcutaneously every 3 weeks. A total of 128 (83.7%) and 68 (89.5%) patients receiving luspatercept and placebo respectively completed 24 weeks of treatment. A total of 78 (51%) and 12 (15.8%) patients receiving luspatercept and placebo respectively completed 48 weeks of treatment. Dose titration up to 1.75 mg/kg was allowed. Dose could be delayed or reduced depending upon Hb level. All patients were eligible to receive best supportive care (BSC), which included RBC transfusions, iron-chelating agents, use of antibiotic, antiviral and antifungal therapy, and nutritional support, as needed. The key baseline disease characteristics in patients with MDS in ACE-536-MDS-001 are shown in Table 4.

Table 4. Baseline characteristics in MDS patients with <5% marrow blasts in ACE-536-MDS001:

 Luspatercept (Ν=153) Placebo (Ν=76)
Demographics
Agea (years)
Median (min, max) 71 (40, 95) 72 (26, 91)
Age categories, n (%)
<64 years
65-74 years
≥75
29 (19.0)
72 (47.1)
52 (34.0)
16 (21.1)
29 (38.2)
31 (40.8)
Sex, n (%)
Male
Female
94 (61.4)
59 (38.6)
50 (65.8)
26 (34.2)
Race, n (%)
Black
White
Not collected or reported
Other
1 (0.7)
107 (69.9)
44 (28.8)
1 (0.7)
0 (0.0)
51 (67.1)
24 (31.6)
1 (1.3)
Disease characteristics
Serum EPO (U/L) categoriesb, n (%)
<200
200 to 500
>500
Missing
88 (57.5)
43 (28.1)
21 (13.7)
1 (0.7)
50 (65.8)
15 (19.7)
11 (14.5)
0
Serum ferritin (µg/L)
Median (min, max) 1089.2 (64, 5968) 1122.1 (165, 5849)
IPSS-R classification risk category, n (%)
Very low
Low
Intermediate
Other
18 (11.8)
109 (71.2)
25 (16.3)
1 (0.7)
6 (7.9)
57 (75.0)
13 (17.1)
0
Baseline RBC Transfusion burden/8 weekc, n (%)
≥6 units

≥6 and <8 units
≥8 and <12 units
≥12 units
<6 units
≥4 and <6 units
<4 units
66 (43.1)

35 (22.9)
24 (15.7)
7 (4.6)
87 (56.9)
41 (26.8)
46 (30.1)
33 (43.4)

15 (20.2)
17 (22.4)
1 (1.3)
43 (56.6)
23 (30.3)
20 (26.3)
Haemoglobind (g/dL)
Median (min, max) 7.6 (6, 10) 7.6 (5, 9)
SF3B1, n (%)
Mutated
Unmutated
Missing
149 (92.2)
12 (7.8)
0
65 (85.5)
10 (13.2)
1 (1.3)

EPO=erythropoietin; IPSS-R=International Prognostic Scoring System-Revised
a Time since original MDS diagnosis was defined as the number of years from the date of original diagnosis to the date of informed consent.
b Baseline EPO was defined as the highest EPO value within 35 days of the first dose of study drug.
c Collected over 16 weeks prior to randomisation. d Baseline haemoglobin was defined as the last value measured on or before the date of the first dose of investigational product (IP). After applying the 14/3 day rule, baseline Hb was defined as the lowest Hb value that was within 35 days on
or prior to the first dose of IP.

The efficacy results are summarised below.

Table 5. Efficacy results in patients with MDS in ACE-536-MDS-001:

Endpoint Luspatercept (N=153) Placebo (Ν=76)
Primary endpoint
RBC-TI ≥8 weeks (Week 1-24)
Number of responders (response rate %)
58 (37.9) 10 (13.2)
Common risk difference on response rate (95% CI) 24.56 (14.48, 34.64)
Odds ratio (95% CI)a 5.065 (2.278, 11.259)
p-valuea<0.0001
Secondary endpoints
RBC-TI ≥12 weeks (Weeks 1-24)
Number of responders (response rate %)
43 (28.1) 6 (7.9)
Common risk difference on response rate (95% CI) 20.00 (10.92, 29.08)
Odds ratio (95% CI)a 5.071 (2.002, 12.844)
p-valuea 0.0002
RBC-TI ≥12 weeks (Weeks 1-48)
Number of responders (response rate %)b
51 (33.3) 9 (11.8)
Common risk difference on response rate (95% CI) 21.37 (11.23, 31.51)
Odds ratio (95% CI)a 4.045 (1.827, 8.956)
p-valuea 0.0003
Transfusion event frequencyc
Weeks 1-24
Interval transfusion rate (95% CI)
6.26 (5.56, 7.05) 9.20 (7.98, 10.60)
Relative risk versus placebo 0.68 (0.58, 0.80)
Weeks 25-48
Interval transfusion rate (95% CI)
6.27 (5.47, 7.19) 8.72 (7.40, 10.28)
Relative risk versus placebo 0.72 (0.60, 0.86)
RBC Transfusion unitsc
Weeks 1-24
Baseline transfusion burden <6 units/8 weeks
LS Mean (SE)
95% CI for LS mean
7.2 (0.58)
6.0, 8.3
12.8 (0.82)
11.1, 14.4
LS mean difference (SE) (luspatercept versus placebo)
95% CI for LS mean difference
-5.6 (1.01)
-7.6, -3.6
Baseline transfusion burden ≥6 units/8 weeks
LS Mean (SE)
95% CI for LS mean
18.9 (0.93)
17.1, 20.8
23.7 (1.32)
21.1, 26.4
LS mean difference (SE) (luspatercept versus placebo)
95% CI for LS mean difference
-4.8 (1.62)
-8.0, -1.6
Weeks 25-48
Baseline transfusion burden <6 units/8 weeks
LS Mean (SE)
95% CI for LS mean
7.5 (0.57)
6.3, 8.6
11.8 (0.82)
10.1, 13.4
LS mean difference (SE) (luspatercept versus placebo)
95% CI for LS mean
-4.3 (1.00)
-6.3, -2.3
Baseline transfusion burden ≥6 units/8 weeks
LS Mean (SE)
95% CI for LS mean
19.6 (1.13)
17.4, 21.9
22.9 (1.60)
19.7, 26.0
LS mean difference (SE) (luspatercept versus placebo) )
95% CI for LS mean difference
-3.3 (1.96)
-7.1, 0.6

RBC-TI: RBC Transfusion Independent; CI: confidence interval
a Cochran-Mantel-Haenszel test stratified for average baseline transfusion burden (≥6 units versus <6 units per 8 weeks), and baseline IPSS-R score (very low or low versus intermediate).
b After the Week 25 disease assessment visit, patients who were no longer deriving benefit discontinued therapy; few placebo patients contributed data for evaluation at the later timepoint compared with luspatercept (n=12 vs. n=78 respectively).
c Post-hoc analysis using baseline imputation.

A treatment effect in favour of luspatercept over placebo was observed in most subgroups analysed using transfusion independence ≥12 weeks (during week 1 to week 24), including patients with high baseline endogenous EPO level (200-500 U/L) (23.3% versus 0%, explorative analysis).

Only limited data are available for the group with transfusion burden of ≥8 units/8 weeks. Safety and efficacy have not been established in patients with a transfusion burden of >12 units/8 weeks.

Exploratory findings

Table 6. Exploratory efficacy results in patients with MDS in ACE-536-MDS-001:

Endpoint Luspatercept (N=153) Placebo (N=76)
mHI-Ea
Weeks 1-24
Number of responders (response rate %)
81 (52.9) 9 (11.8)
(95% CI) (44.72, 61.05) (5.56, 21.29)
RBC transfusion reduction of 4 units/8 weeks, n (%) 52/107 (48.6) 8/56 (14.3)
Mean haemoglobin increase of ≥1.5 g/dL for 8 weeks, n (%) 29/46 (63.0) 1/20 (5.0)
Weeks 1-48
Number of responders (response rate %)
90 (58.8) 13 (17.1)
(95% CI) (50.59, 66.71) (9.43, 27.47)
RBC transfusion reduction of 4 units/8 weeks, n (%) 58/107 (54.2) 12/56 (21.4)
Mean haemoglobin increase of ≥1.5 g/dL for 8 weeks, n (%) 32/46 (69.6) 1/20 (5.0)
Mean change from baseline in mean serum ferritin with imputation by baseline (ITT population)
Mean change from baseline in mean serum ferritin averaged over Weeks 9 through 24 (μg/L)b
LS Mean (SE)
9.9 (47.09) 190.0 (60.30)
95% CI for LS mean -82.9, 102.7 71.2, 308.8
Treatment Comparison (Luspatercept vs Placebo)c
LS Mean Difference (SE) -180.1 (65.81)
95% CI for LS Mean Difference -309.8, -50.4

a mHI-E = modified haematological improvement – erythroid. The proportion of patients meeting the HI-E criteria as per International Working Group (IWG) 2006 criteria sustained over a consecutive 56-day period during the indicated treatment period. For patients with baseline RBC transfusion burden of ≥4 units/8 weeks, mHI-E was defined as a reduction in RBC transfusion of at least 4 units/8 weeks. For patients with baseline RBC transfusion burden of <4 units/8 weeks, mHI-E was
defined as a mean increase in Hb of ≥1.5 g/dL for 8 weeks in the absence of RBC transfusions.
b If a subject did not have a serum ferritin value within the designated postbaseline interval, the serum ferritin is imputed from the baseline value.
c Analysis of covariance was used to compare the treatment difference between groups (including nominal p-value), with the change in serum ferritin as the dependent variable, treatment group (2 levels) as a factor, and baseline serum ferritin value as covariates, stratified by average baseline RBC transfusion requirement (≥6 units versus <6 units of RBC per 8 weeks), and baseline IPSS-R (very low or low versus intermediate).

The median duration of the longest RBC Transfusion Independent (RBC-TI) period among responders in the luspatercept treatment arm was 30.6 weeks.

62.1% (36/58) of the luspatercept responders who achieved RBC-TI ≥ 8 weeks from Week 1-24 had 2 or more episodes of RBC-TI at the time of analysis.

β-thalassaemia

The efficacy and safety of luspatercept were evaluated in a Phase 3 multicentre, randomised, double-blind, placebo-controlled study BELIEVE (ACE-536-B-THAL-001) in adult patients with β-thalassaemia–associated anaemia who require RBC transfusions (6-20 RBC units/24 weeks) with no transfusion-free period >35 days during that period.

Patients in both the luspatercept and placebo arms were treated for at least 48 and up to 96 weeks. After unblinding, placebo patients were able to cross-over to luspatercept.

A total of 336 adult patients were randomised to receive luspatercept 1.0 mg/kg (n=224) or placebo (n=112) subcutaneously every 3 weeks. Dose titration to 1.25 mg/kg was allowed. Dose could be delayed or reduced depending upon Hb level. All patients were eligible to receive BSC, which included RBC transfusions, iron-chelating agents, use of antibiotic, antiviral and antifungal therapy, and nutritional support, as needed. The study excluded patients with haemoglobin S/β-thalassaemia or alpha (α)-thalassaemia or who had major organ damage (liver disease, heart disease, lung disease, renal insufficiency). Patients with recent DVT or stroke or recent use of ESA, immunosuppressant or hydroxyurea therapy were also excluded. The key baseline disease characteristics in patients with β-thalassaemia in ACE-536-B-THAL-001 are shown in Table 7.

Table 7. Baseline characteristics in patients with β-thalassaemia in ACE-536-B-THAL-001:

 Luspatercept (Ν=224) Placebo (Ν=112)
Demographics
Age (years)
Median (min, max) 30.0 (18, 66) 30.0 (18, 59)
Age categories, n (%)
≤32
>32 to ≤50
>50
129 (57.6)
78 (34.8)
17 (7.6)
63 (56.3)
44 (39.3)
5 (4.5)
Sex, n (%)
Male
Female
92 (41.1)
132 (58.9)
49 (43.8)
63 (56.3)
Race, n (%)
Asian
Black
White
Not collected or reported
Other
81 (36.2)
1 (0.4)
122 (54.5)
5 (2.2)
15 (6.7)
36 (32.1)
0
60 (53.6)
5 (4.5)
11 (9.8)
Disease characteristics
Pretransfusion Hb thresholda, 12 week run-in (g/dL)
Median (min, max) 9.30 (4.6, 11.4) 9.16 (6.2, 11.5)
Baseline transfusion burden 12 weeks
Median (min, max) (units/12 weeks) (Week -12 to Day 1) 6.12 (3.0, 14.0) 6.27 (3.0, 12.0)
β-thalassaemia gene mutation grouping, n (%)
β0/β0
Non-β0/β0
Missingb
68 (30.4)
155 (69.2)
1 (0.4)
35 (31.3)
77 (68.8)
0

a The 12-week pretransfusion threshold was defined as the mean of all documented pretransfusions hb values for a subject during the 12 weeks prior to Cycle 1 Day 1.
b “Missing” category includes patients in the population who had no result for the parameter listed.

The study was unblinded for analyses when all patients had at least received 48 weeks of treatment or discontinued treatment.

The efficacy results are summarised below.

Table 8. Efficacy results in patients with β-thalassaemia in ACE-536-B-THAL-001:

Endpoint Luspatercept (Ν=224) Placebo (Ν=112)
≥33% reduction from baseline in RBC transfusion burden with a reduction of at least 2 units for 12 consecutive weeks compared to the 12-week interval prior to treatment
Primary endpoint – Weeks 13-24 48 (21.4) 5 (4.5)
Difference in proportions (95% CI)a 17.0 (10.4, 23.6)
p-valueb <0.0001
Weeks 37-48 44 (19.6) 4 (3.6)
Difference in proportions (95% CI)a 16.1 (9.8, 22.3)
p-valueb <0.0001
≥50% reduction from baseline in RBC transfusion burden with a reduction of at least 2 units for 12 consecutive weeks compared to the 12-week interval prior to treatment
Weeks 13-24 17 (7.6) 2 (1.8)
Difference in proportions (95% CI)sup>a 5.8 (1.6, 10.1)
p-valueb0.0303
Weeks 37-48 23 (10.3) 1 (0.9)
Difference in proportions (95% CI)a 9.4 (5.0, 13.7)
p-valueb0.0017

CI: confidence interval.
a Difference in proportions (luspatercept + BSC – placebo + BSC) and 95% CIs estimated from the unconditional exact test.
b P-value from the Cochran Mantel-Haenszel test stratified by the geographical region.

Exploratory findings

Table 9. Exploratory efficacy results in patients with β-thalassaemia in ACE-536-B-THAL-001:

Endpoint Luspatercept (N=224) Placebo (N=112)
≥33% reduction from baseline in RBC transfusion burden with a reduction of at least 2 units for 12 consecutive weeks compared to the 12-week interval prior to treatment
Any consecutive 12 weeks* 158 (70.5) 33 (29.5)
Difference in proportions (95% CI)a 41.1 (30.7, 51.4)
Any consecutive 24 weeks* 92 (41.1) 3 (2.7)
Difference in proportions (95% CI)a 38.4 (31.3, 45.5)
≥50% reduction from baseline in RBC transfusion burden with a reduction of at least 2 units for 12 consecutive weeks compared to the 12-week interval prior to treatment
Any consecutive 12 weeks* 90 (40.2) 7 (6.3)
Difference in proportions (95% CI)a 33.9 (26.1, 41.8)
Any consecutive 24 weeks* 37 (16.5) 1 (0.9)
Difference in proportions (95% CI)a 15.6 (10.5, 20.8)
Least square (LS) mean change from baseline in transfusion burden (RBC units/48 weeks)
Weeks 1 to Week 48
LS mean
-4.67 +1.16
LS mean of difference (luspatercept-placebo) (95% CI)b -5.83 (-7.01, -4.6)
Weeks 49 to Week 96
LS mean
-5.66 +2.19
LS mean of difference (luspatercept-placebo) (95% CI)b -7.84 (-14.44, -1.25)

CI: confidence interval.
a Difference in proportions (luspatercept + BSC – placebo + BSC) and 95% CIs estimated from the unconditional exact test.
b Estimates are based on ANCOVA model with geographical regions and baseline transfusion burden as covariates

A reduction in mean serum ferritin levels was observed from baseline in the luspatercept arm compared to an increase in the placebo arm at Week 48 (-233.51 μg/L versus +114.28 μg/L which resulted in a least square mean treatment difference of -347.8 µg/L (95% CI: -516.95, -178.65).

80.4% (127/158) of luspatercept responders who achieved at least a 33% reduction in transfusion burden during any consecutive 12-week interval achieved 2 or more episodes of response at the time of analysis.

Paediatric population

Myelodysplastic syndromes

The European Medicines Agency has waived the obligation to submit the results of studies with Reblozyl in all subsets of the paediatric population in myelodysplastic syndromes (see section 4.2 for information on paediatric use).

β-thalassaemia

The European Medicines Agency has deferred the obligation to submit the results of studies with Reblozyl in one or more subsets of paediatric population older than 6 months of age in β-thalassaemia (see section 4.2 for information on paediatric use).

5.2. Pharmacokinetic properties

Absorption

In healthy volunteers and patients, luspatercept is slowly absorbed following subcutaneous administration, with the Cmax in serum often observed approximately 7 days post-dose across all dose levels. Population pharmacokinetic (PK) analysis suggests that the absorption of luspatercept into the circulation is linear over the range of studied doses, and the absorption is not significantly affected by the subcutaneous injection location (upper arm, thigh or abdomen). Interindividual variability in AUC was approximately 38% in MDS patients and 36% in β-thalassaemia patients.

Distribution

At the recommended doses, the mean apparent volume of distribution was 9.68 L for MDS patients and 7.08 L for β-thalassaemia patients. The small volume of distribution indicates that luspatercept is confined primarily in extracellular fluids, consistent with its large molecular mass.

Biotransformation

Luspatercept is expected to be catabolised into amino acids by general protein degradation process.

Elimination

Luspatercept is not expected to be excreted into urine due to its large molecular mass that is above the glomerular filtration size exclusion threshold. At the recommended doses, the mean apparent total clearance was 0.516 L/day for MDS patients and 0.437 L/day for β-thalassaemia. The mean half-life in serum was approximately 13 days for MDS patients and 11 days for β-thalassaemia patients.

Linearity/non-linearity

The increase of luspatercept Cmax and AUC in serum is approximately proportional to increases in dose from 0.125 to 1.75 mg/kg. Luspatercept clearance was independent of dose or time.

When administered every three weeks, luspatercept serum concentration reaches the steady state after 3 doses, with an accumulation ratio of approximately 1.5.

Haemoglobin response

In patients who received <4 units of RBC transfusion within 8 weeks prior to the study, Hb increased within 7 days of treatment initiation and the increase correlated with the time to reach luspatercept Cmax. The greatest mean Hb increase was observed after the first dose, with additional smaller increases observed after subsequent doses. Hb levels returned to baseline value approximately 6 to 8 weeks from the last dose (0.6 to 1.75 mg/kg). Increasing luspatercept serum exposure (AUC) was associated with a greater Hb increase in patients with MDS or β-thalassaemia.

Special populations

Elderly

Population PK analysis for luspatercept included patients with ages ranging from 18 to 95 years old, with a median age of 72 years for MDS patients and of 32 years for β-thalassaemia patients. No clinically significant difference in AUC or clearance was found across age groups (<65, 65-74, and ≥75 years for MDS patients; 18-23, 24-31, 32-41, and 42-66 years for β-thalassaemia patients).

Hepatic impairment

Population PK analysis for luspatercept included patients with normal hepatic function (BIL, ALT, and AST ≤ ULN; N=207), mild hepatic impairment (BIL >1–1.5 x ULN, and ALT or AST > ULN; N=160), moderate hepatic impairment (BIL >1.5–3 x ULN, any ALT or AST; N=138), or severe hepatic impairment (BIL >3 x ULN, any ALT or AST; N=40) as defined by the National Cancer Institute criteria of hepatic dysfunction. Effects of hepatic function categories, elevated liver enzymes (ALT or AST, up to 3 x ULN) and elevated total BIL (4–246 mol/L) on luspatercept clearance were not observed. No clinically significant difference in mean steady state Cmax and AUC was found across hepatic function groups. PK data are insufficient for patients with liver enzymes (ALT or AST) ≥3 x ULN.

Renal impairment

Population PK analysis for luspatercept included patients with normal renal function (eGFR ≥90 mL/min/1.73 m²; N=315), mild renal impairment (eGFR 60 to 89 mL/min/1.73 m²; N=171), or moderate renal impairment (eGFR 30 to 59 mL/min/1.73 m²; N=59). No clinically significant difference in mean steady state Cmax and AUC was found across renal function groups. PK data are not available for patients with severe renal impairment (eGFR <30 mL/min/1.73 m²) or end-stage kidney disease.

Other intrinsic factors

The following population characteristics have no clinically significant effect on luspatercept AUC or clearance: sex and race (Asian versus White).

The following baseline disease characteristics had no clinically significant effect on luspatercept clearance: serum erythropoietin level, RBC transfusion burden, MDS ring sideroblasts, β-thalassaemia genotype (β0/β0 versus non-β0/β0) and splenectomy.

The volume of distribution and clearance of luspatercept increased with increase of body weight, supporting the body weight-based dosing regimen.

5.3. Preclinical safety data

Single and repeat-dose toxicity

Following repeated administration of luspatercept in rats, toxicities included: membranoproliferative glomerulonephritis; congestion, necrosis and/or mineralisation of the adrenal glands; hepatocellular vacuolation and necrosis; mineralisation of the glandular stomach; and decreased heart and lung weights with no associated histology findings. A clinical observation of swollen hindlimbs/feet was noted in several studies in rats and rabbits (including juvenile and reproductive toxicity studies). In one juvenile rat, this correlated histopathologically with new bone formation, fibrosis, and inflammation. Membranoproliferative glomerulonephritis was also seen in monkeys. Additional toxicities in monkeys included: vascular degeneration and inflammatory infiltrates in the choroid plexus.

For the 6-month toxicity study, the longest duration study in monkeys, the no-observed-adverse-effect level (NOAEL) was 0.3 mg/kg (0.3-fold of clinical exposure at 1.75 mg/kg every 3 weeks). A NOAEL was not identified in rats and the lowest-observed-adverse-effect-level (LOAEL) in the rat 3-month study was 1 mg/kg (0.9-fold of clinical exposure at 1.75 mg/kg every 3 weeks).

Carcinogenesis and mutagenesis

Neither carcinogenicity nor mutagenicity studies with luspatercept have been conducted. Haematological malignancies were observed in 3 out of 44 rats examined in the highest dose group (10 mg/kg) in the definitive juvenile toxicity study. The occurrence of these tumours in young animals is unusual and the relationship to luspatercept therapy cannot be ruled out. At the 10 mg/kg dose, at which tumours were observed, the exposure represents an exposure multiple of approximately 4 times the estimated exposure at a clinical dose of 1.75 mg/kg every three weeks.

No other proliferative or pre-neoplastic lesions, attributable to luspatercept, have been observed in any species in other non-clinical safety studies conducted with luspatercept, including the 6-month study in monkeys.

Fertility

In a fertility study in rats, administration of luspatercept to females at doses higher than the currently recommended highest human dose reduced the average number of corpora lutea, implantations and viable embryos. No such effects were observed when exposure in animals was at 1.5 times the clinical exposure. Effects on fertility in female rats were reversible after a 14-week recovery period.

Administration of luspatercept to male rats at doses higher than the currently recommended highest human dose had no adverse effect on male reproductive organs or on their ability to mate and produce viable embryos. The highest dose tested in male rats yielded an exposure approximately 7 times the clinical exposure.

Embryo-foetal development (EFD)

Embryo-foetal developmental toxicology studies (range-finding and definitive studies) were conducted in pregnant rats and rabbits. In the definitive studies, doses of up to 30 mg/kg or 40 mg/kg every week were administered twice during the period of organogenesis. Luspatercept was a selective developmental toxicant (dam not affected; foetus affected) in the rat and a maternal and foetal developmental toxicant (doe and foetus affected) in the rabbit. Embryofoetal effects were seen in both species and included reductions in numbers of live foetuses and foetal body weights, increases in resorptions, post-implantation loss and skeletal variations and, in rabbit foetuses, malformations of the ribs and vertebrae. In both species, effects of luspatercept were observed in the EFD studies at the lowest dose tested, 5 mg/kg, which corresponds to an estimated exposure in rats and rabbits of approximately 2.7 and 5.5 times greater, respectively, than the estimated clinical exposure.

Pre- and post-natal development

In a pre- and post-natal development study, with dose levels of 3, 10, or 30 mg/kg administered once every 2 weeks from gestational day (GD) 6 through post-natal day (PND) 20, adverse findings at all doses consisted of lower F1 pup body weights in both sexes at birth, throughout lactation, and post weaning (PND 28); lower body weights during the early premating period (Week 1 and 2) in the F1 females (adverse only at the 30 mg/kg/dose) and lower body weights in F1 males during the premating, pairing and post-mating periods; and microscopic kidney findings in F1 pups. Additionally, non-adverse findings included delayed male sexual maturation at the 10 and 30 mg/kg/dose. The delay in growth and the adverse kidney findings, in the F1 generation, precluded the determination of a NOAEL for F1 general and developmental toxicity. However, there was no effect on behavioural indices, fertility or reproductive parameters at any dose level in either sex, therefore the NOAEL for behavioural assessments, fertility and reproductive function in the F1 animals was considered to be the 30 mg/kg/dose. Luspatercept is transferred through the placenta of pregnant rats and rabbits and is excreted into the milk of lactating rats.

Juvenile toxicity

In a study in juvenile rats, luspatercept was administered from postnatal day (PND) 7 to PND 91 at 0, 1, 3, or 10 mg/kg. Many of the findings seen in repeat-dose toxicity studies in adult rats were repeated in the juvenile rats. These findings included glomerulonephritis in the kidney, haemorrhage/congestion, necrosis and mineralization of the adrenal gland, mucosal mineralization in the stomach, lower heart weights, and swollen hindlimbs/feet. Luspatercept-related findings unique to juvenile rats included tubular atrophy/hypoplasia of the kidney inner medulla, delays in the mean age of sexual maturation in males, effects on reproductive performance (lower mating indices), and nonadverse decreases in bone mineral density in both male and female rats. The effects on reproductive performance were observed after a greater than 3-month recovery period, suggesting a permanent effect. Although reversibility of the tubular atrophy/hypoplasia was not examined, these effects are also considered to be irreversible. Adverse effects on the kidney and reproductive system were observed at clinically relevant exposure levels and seen at the lowest dose tested and, thus, an NOAEL was not established. In addition, haematological malignancies were observed in 3 out of 44 rats examined in the highest dose group (10 mg/kg). These findings are all considered potential risks in paediatric patients.

© All content on this website, including data entry, data processing, decision support tools, "RxReasoner" logo and graphics, is the intellectual property of RxReasoner and is protected by copyright laws. Unauthorized reproduction or distribution of any part of this content without explicit written permission from RxReasoner is strictly prohibited. Any third-party content used on this site is acknowledged and utilized under fair use principles.